Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Intervalo de año de publicación
1.
Gene Ther ; 24(8): 462-469, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28617420

RESUMEN

The production of high-titer recombinant adeno-associated virus (rAAV) vector is essential for treatment of genetic diseases affecting the retina and choroid, where anatomical constraints may limit injectable volumes. Problematically, cytotoxicity arising from overexpression of the transgene during vector production frequently leads to a reduction in vector yield. Herein, we evaluate the use of microRNA (miRNA)-mediated silencing to limit overexpression of cytotoxic transgenes during packaging as a method of increasing vector yield. We examined if post-transcriptional regulation of transgenes during packaging via miRNA technology would lead to increased rAAV yields. Our results demonstrate that silencing of cytotoxic transgenes during production resulted in up to a 22-fold increase in vector yield. The inclusion of organ-specific miRNA sequences improved biosafety by limiting off-target expression following systemic rAAV administration. The small size (22-23 bp) of the target site allows for the inclusion of multiple copies into the vector with minimal impact on coding capacity. Taken together, our results suggest that inclusion of miRNA target sites into the 3'-untranslated region of the AAV cassette allow for silencing of cytotoxic transgenes during vector production leading to improved vector yield, in addition to increasing targeting specificity without reliance on cell-specific promoters.


Asunto(s)
Dependovirus/genética , Silenciador del Gen , ARN Interferente Pequeño/administración & dosificación , Tratamiento con ARN de Interferencia/métodos , Replicación Viral , Animales , Dependovirus/fisiología , Marcación de Gen/métodos , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/efectos adversos , ARN Interferente Pequeño/genética , Transgenes/genética
2.
Gene Ther ; 23(2): 223-30, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26467396

RESUMEN

Delivery of therapeutic transgenes to retinal photoreceptors using adeno-associated virus (AAV) vectors has traditionally required subretinal injection. Recently, photoreceptor transduction efficiency following intravitreal injection (IVT) has improved in rodent models through use of capsid-mutant AAV vectors; but remains limited in large animal models. Thickness of the inner limiting membrane (ILM) in large animals is thought to impair retinal penetration by AAV. Our study compared two newly developed AAV vectors containing multiple capsid amino acid substitutions following IVT in dogs. The ability of two promoter constructs to restrict reporter transgene expression to photoreceptors was also evaluated. AAV vectors containing the interphotoreceptor-binding protein (IRBP) promoter drove expression exclusively in rod and cone photoreceptors, with transduction efficiencies of ~4% of cones and 2% of rods. Notably, in the central region containing the cone-rich visual streak, 15.6% of cones were transduced. Significant regional variation existed, with lower transduction efficiencies in the temporal regions of all eyes. This variation did not correlate with ILM thickness. Vectors carrying a cone-specific promoter failed to transduce a quantifiable percentage of cone photoreceptors. The newly developed AAV vectors containing the IRBP promoter were capable of producing photoreceptor-specific transgene expression following IVT in the dog.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Células Fotorreceptoras de Vertebrados/metabolismo , Animales , Perros , Elementos de Facilitación Genéticos , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Inyecciones Intravítreas , Regiones Promotoras Genéticas , Retina/metabolismo , Transducción Genética
3.
Gene Ther ; 23(3): 272-82, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26704722

RESUMEN

Direct gene delivery to the neurons of interest, without affecting other neuron populations in the cerebral cortex, represent a challenge owing to the heterogeneity and cellular complexity of the brain. Genetic modulation of corticospinal motor neurons (CSMN) is required for developing effective and long-term treatment strategies for motor neuron diseases, in which voluntary movement is impaired. Adeno-associated viruses (AAV) have been widely used for neuronal transduction studies owing to long-term and stable gene expression as well as low immunoreactivity in humans. Here we report that AAV2-2 transduces CSMN with high efficiency upon direct cortex injection and that transduction efficiencies are similar during presymptomatic and symptomatic stages in hSOD1(G93A) transgenic amyotrophic lateral sclerosis (ALS) mice. Our findings reveal that choice of promoter improves selectivity as AAV2-2 chicken ß-actin promoter injection results in about 70% CSMN transduction, the highest percentage reported to date. CSMN transduction in both wild-type and transgenic ALS mice allows detailed analysis of single axon fibers within the corticospinal tract in both cervical and lumbar spinal cord and reveals circuitry defects, which mainly occur between CSMN and spinal motor neurons in hSOD1(G93A) transgenic ALS mice. Our findings set the stage for CSMN gene therapy in ALS and related motor neuron diseases.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/uso terapéutico , Corteza Motora/metabolismo , Enfermedad de la Neurona Motora/genética , Enfermedad de la Neurona Motora/terapia , Transducción Genética , Animales , Animales Modificados Genéticamente , Terapia Genética , Ratones , Neuronas Motoras/metabolismo
4.
Gene Ther ; 23(6): 548-56, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27052802

RESUMEN

Adeno-associated virus (AAV) vector-based gene therapy is a promising treatment strategy for delivery of neurotrophic transgenes to retinal ganglion cells (RGCs) in glaucoma patients. Retinal distribution of transgene expression following intravitreal injection (IVT) of AAV is variable in animal models and the vitreous humor may represent a barrier to initial vector penetration. The primary goal of our study was to investigate the effect of prior core vitrectomy with posterior hyaloid membrane peeling on pattern and efficiency of transduction of a capsid amino acid substituted AAV2 vector, carrying the green fluorescent protein (GFP) reporter transgene following IVT in dogs. When progressive intraocular inflammation developed starting 4 weeks post IVT, the study plan was modified to allow detailed characterization of the etiology as a secondary goal. Unexpectedly, surgical vitrectomy was found to significantly limit transduction, whereas in non-vitrectomized eyes transduction efficiency reached upwards to 37.3% of RGC layer cells. The developing retinitis was characterized by mononuclear cell infiltrates resulting from a delayed-type hypersensitivity reaction, which we suspect was directed at the GFP transgene. Our results, in a canine large animal model, support caution when considering surgical vitrectomy before IVT for retinal gene therapy in patients, as prior vitrectomy appears to significantly reduce transduction efficiency and may predispose the patient to development of vector-induced immune reactions.


Asunto(s)
Dependovirus/genética , Vitrectomía , Animales , Perros , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Humanos , Retina/metabolismo , Transducción Genética , Transgenes
5.
Gene Ther ; 22(2): 138-45, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25427613

RESUMEN

We evaluated the effect of AAV2- and 17-AAG (17-N-allylamino-17-demethoxygeldanamycin)-mediated upregulation of Hsp70 expression on the survival of retinal ganglion cells (RGCs) injured by optic nerve crush (ONC). AAV2-Hsp70 expression in the retina was primarily observed in the ganglion cell layer. Approximately 75% of all transfected cells were RGCs. RGC survival in AAV2-Hsp70-injected animals was increased by an average of 110% 2 weeks after the axonal injury compared with the control. The increase in cell numbers was not even across the retinas with a maximum effect of approximately 306% observed in the inferior quadrant. 17-AAG-mediated induction of Hsp70 expression has been associated with cell protection in various models of neurodegenerative diseases. We show here that a single intravitreal injection of 17-AAG (0.2 ug ul(-1)) results in an increased survival of ONC-injured RGCs by approximately 49% compared with the vehicle-treated animals. Expression of Hsp70 in retinas of 17-AAG-treated animals was upregulated approximately by twofold compared with control animals. Our data support the idea that the upregulation of Hsp70 has a beneficial effect on the survival of injured RGCs, and the induction of this protein could be viewed as a potential neuroprotective strategy for optic neuropathies.


Asunto(s)
Benzoquinonas/farmacología , Dependovirus/genética , Proteínas HSP70 de Choque Térmico/genética , Lactamas Macrocíclicas/farmacología , Traumatismos del Nervio Óptico/terapia , Células Ganglionares de la Retina/fisiología , Animales , Axones/patología , Supervivencia Celular , Terapia Combinada , Terapia Genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Ratones Endogámicos C57BL , Compresión Nerviosa , Regeneración Nerviosa , Retina/metabolismo , Retina/patología , Activación Transcripcional , Transducción Genética
6.
Gene Ther ; 22(8): 619-27, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25965394

RESUMEN

Loss of SPATA7 function causes the pathogenesis of Leber congenital amaurosis and retinitis pigmentosa. Spata7 knockout mice mimic human SPATA7-related retinal disease with apparent photoreceptor degeneration observed as early as postnatal day 15 (P15). To test the efficacy of adeno-associated virus (AAV)-mediated gene therapy for rescue of photoreceptor survival and function in Spata7 mutant mice, we employed the AAV8(Y733F) vector carrying hGRK1-driven full-length FLAG-tagged Spata7 cDNA to target both rod and cone photoreceptors. Following subretinal injection of this vector, FLAG-tagged SPATA7 was found to colocalize with endogenous SPATA7 in wild-type mice. In Spata7 mutant mice initially treated at P15, we observed improvement of photoresponse, photoreceptor ultrastructure and significant alleviation of photoreceptor degeneration. Furthermore, we performed treatments at P28 and P56 and found that all treatments (P15-P56) can ameliorate rod and cone loss in the long term (1 year); however, none efficiently protect photoreceptors from degeneration by 86 weeks of age as only a small amount of treated photoreceptors can survive to this time. This study demonstrates long-term improvement of photoreceptor function by AAV8(Y733F)-introduced Spata7 expression in a mouse model as potential treatment of the human disease, but also suggests that treated mutant photoreceptors still undergo progressive degeneration.


Asunto(s)
Proteínas de Unión al ADN/genética , Dependovirus/genética , Terapia Genética , Amaurosis Congénita de Leber/terapia , Retinitis Pigmentosa/terapia , Animales , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Vectores Genéticos , Amaurosis Congénita de Leber/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/genética
7.
Gene Ther ; 21(1): 96-105, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24225638

RESUMEN

Recombinant adeno-associated viruses are important vectors for retinal gene delivery. Currently utilized vectors have relatively slow onset, and for efficient transduction it is necessary to deliver treatment subretinally, with the potential for damage to the retina. Amino-acid substitutions in the viral capsid improve efficiency in rodent eyes by evading host responses. As dogs are important large animal models for human retinitis pigmentosa, we evaluated the speed and efficiency of retinal transduction using capsid-mutant vectors injected both subretinally and intravitreally. We evaluated AAV serotypes 2 and 8 with amino-acid substitutions of surface-exposed capsid tyrosine residues. The chicken beta-actin promoter was used to drive green fluorescent protein expression. Twelve normal adult beagles were injected; four dogs received intravitreal injections and eight dogs received subretinal injections. Capsid-mutant viruses tested included AAV2(quad Y-F) (intravitreal and subretinal) and self-complementary scAAV8(Y733F) (subretinal only). Contralateral control eyes received injections of scAAV5 (subretinal) or scAAV2 (intravitreal). Subretinally delivered vectors had a faster expression onset than intravitreally delivered vectors. Subretinally delivered scAAV8(Y733F) had a faster onset of expression than scAAV5. All subretinally injected vector types transduced the outer retina with high efficiency and the inner retina with moderate efficiency. Intravitreally delivered AAV2(quad Y-F) had a marginally higher efficiency of transduction of both outer retinal and inner retinal cells than scAAV2. Because of their rapid expression onset and efficient transduction, subretinally delivered capsid-mutant AAV8 vectors may increase the efficacy of gene therapy treatment for rapid photoreceptor degenerative diseases. With further refinement, capsid-mutant AAV2 vectors show promise for retinal gene delivery from an intravitreal approach.


Asunto(s)
Cápside , Dependovirus/genética , Vectores Genéticos , Retina/metabolismo , Sustitución de Aminoácidos , Animales , Dependovirus/fisiología , Perros , Femenino , Humanos , Inyecciones Intraoculares , Masculino , Mutación , Proteínas Recombinantes/metabolismo , Retina/virología , Transducción Genética , Tirosina , Tropismo Viral
8.
Gene Ther ; 20(8): 824-33, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23344065

RESUMEN

Usher 1 patients are born profoundly deaf and then develop retinal degeneration. Thus they are readily identified before the onset of retinal degeneration, making gene therapy a viable strategy to prevent their blindness. Here, we have investigated the use of adeno-associated viruses (AAVs) for the delivery of the Usher 1B gene, MYO7A, to retinal cells in cell culture and in Myo7a-null mice. MYO7A cDNA, under control of a smCBA promoter, was packaged in single AAV2 and AAV5 vectors and as two overlapping halves in dual AAV2 vectors. The 7.9-kb smCBA-MYO7A exceeds the capacity of an AAV vector; packaging of such oversized constructs into single AAV vectors may involve fragmentation of the gene. Nevertheless, the AAV2 and AAV5 single vector preparations successfully transduced photoreceptor and retinal pigment epithelium cells, resulting in functional, full-length MYO7A protein and correction of mutant phenotypes, suggesting successful homologous recombination of gene fragments. With discrete, conventional-sized dual AAV2 vectors, full-length MYO7A was detected, but the level of protein expression was variable, and only a minority of cells showed phenotype correction. Our results show that MYO7A therapy with AAV2 or AAV5 single vectors is efficacious; however, the dual AAV2 approach proved to be less effective.


Asunto(s)
Terapia Genética , Miosinas/genética , Degeneración Retiniana/genética , Degeneración Retiniana/terapia , Síndromes de Usher/terapia , Animales , ADN Complementario , Dependovirus , Expresión Génica , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Miosina VIIa , Miosinas/metabolismo , Retina/patología , Degeneración Retiniana/patología , Síndromes de Usher/genética , Síndromes de Usher/patología
9.
Gene Ther ; 20(6): 670-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23151520

RESUMEN

With the long-term goal of developing a gene-based treatment for osteoarthritis (OA), we performed studies to evaluate the equine joint as a model for adeno-associated virus (AAV)-mediated gene transfer to large, weight-bearing human joints. A self-complementary AAV2 vector containing the coding regions for human interleukin-1-receptor antagonist (hIL-1Ra) or green fluorescent protein was packaged in AAV capsid serotypes 1, 2, 5, 8 and 9. Following infection of human and equine synovial fibroblasts in culture, we found that both were only receptive to transduction with AAV1, 2 and 5. For these serotypes, however, transgene expression from the equine cells was consistently at least 10-fold higher. Analyses of AAV surface receptor molecules and intracellular trafficking of vector genomes implicate enhanced viral uptake by the equine cells. Following delivery of 1 × 10(11) vector genomes of serotypes 2, 5 and 8 into the forelimb joints of the horse, all three enabled hIL-1Ra expression at biologically relevant levels and effectively transduced the same cell types, primarily synovial fibroblasts and, to a lesser degree, chondrocytes in articular cartilage. These results provide optimism that AAV vectors can be effectively adapted for gene delivery to large human joints affected by OA.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Proteína Antagonista del Receptor de Interleucina 1/genética , Osteoartritis/genética , Animales , Cartílago Articular/metabolismo , Cartílago Articular/patología , Cartílago Articular/virología , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Caballos , Humanos , Interleucina-1/genética , Articulaciones/metabolismo , Articulaciones/patología , Articulaciones/virología , Osteoartritis/terapia , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Membrana Sinovial/virología
10.
Nat Genet ; 28(1): 92-5, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11326284

RESUMEN

The relationship between the neurosensory photoreceptors and the adjacent retinal pigment epithelium (RPE) controls not only normal retinal function, but also the pathogenesis of hereditary retinal degenerations. The molecular bases for both primary photoreceptor and RPE diseases that cause blindness have been identified. Gene therapy has been used successfully to slow degeneration in rodent models of primary photoreceptor diseases, but efficacy of gene therapy directed at photoreceptors and RPE in a large-animal model of human disease has not been reported. Here we study one of the most clinically severe retinal degenerations, Leber congenital amaurosis (LCA). LCA causes near total blindness in infancy and can result from mutations in RPE65 (LCA, type II; MIM 180069 and 204100). A naturally occurring animal model, the RPE65-/- dog, suffers from early and severe visual impairment similar to that seen in human LCA. We used a recombinant adeno-associated virus (AAV) carrying wild-type RPE65 (AAV-RPE65) to test the efficacy of gene therapy in this model. Our results indicate that visual function was restored in this large animal model of childhood blindness.


Asunto(s)
Ceguera/terapia , Modelos Animales de Enfermedad , Enfermedades de los Perros/genética , Proteínas del Ojo/genética , Terapia Genética/métodos , Atrofias Ópticas Hereditarias/terapia , Proteínas/genética , Animales , Animales Modificados Genéticamente , Proteínas Portadoras , Dependovirus/genética , Perros , cis-trans-Isomerasas
11.
Gene Ther ; 19(3): 255-63, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21697953

RESUMEN

The auditory portion of the inner ear, the cochlea, is an ideal organ for local gene transfection owing to its relative isolation. Various carriers have been tested for cochlear gene transfection. To date, viral vectors appear to have much higher transfection efficacy than non-viral mechanisms. Among these vectors, recombinant adeno-associated virus (rAAV) vectors have several advantages such as being non-pathogenic and the ability to produce prolonged gene expression in various cell types. However, rAAV vectors cannot pass through the intact round window membrane (RWM), otherwise a very attractive approach to access the human inner ear. In this study, performed in guinea-pigs, we describe a method to increase the permeability of RWM to rAAV vectors by partial digestion with collagenase solution. Elevated delivery of rAAV across the partially digested RWM increased transfection efficacy to a satisfactory level, even though it was still lower than that achieved by direct cochleostomy injection. Functional tests (auditory brainstem responses) showed that this enzymatic manipulation did not cause permanent hearing loss if applied appropriately. Morphological observations suggested that the damage to RWM caused by partial digestion healed within four weeks. Taken together, these findings suggest that partial digestion of the RWM is a safe and effective method for increasing the transfection of cochlear sensory cells with rAAV.


Asunto(s)
Cóclea/metabolismo , Dependovirus/metabolismo , Vectores Genéticos/metabolismo , Ventana Redonda/metabolismo , Transfección , Animales , Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Cobayas , Células Ciliadas Auditivas/metabolismo , Humanos , Masculino
13.
Nat Med ; 4(8): 967-71, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9701253

RESUMEN

Ribozymes, catalytic RNA molecules that cleave a complementary mRNA sequence, have potential as therapeutics for dominantly inherited disease. Twelve percent of American patients with the blinding disease autosomal dominant retinitis pigmentosa (ADRP) carry a substitution of histidine for proline at codon 23 (P23H) in their rhodopsin gene, resulting in photoreceptor cell death from the synthesis of the abnormal gene product. Ribozymes can discriminate and catalyze the in vitro destruction of P23H mutant mRNAs from a transgenic rat model of ADRP. Here, we demonstrate that in vivo expression of either a hammerhead or hairpin ribozyme in this rat model considerably slows the rate of photoreceptor degeneration for at least three months. Catalytically inactive control ribozymes had less effect on the retinal degeneration. Intracellular production of ribozymes in photoreceptors was achieved by transduction with a recombinant adeno-associated virus (rAAV) incorporating a rod opsin promoter. Ribozyme-directed cleavage of mutant mRNAs, therefore, may be an effective therapy for ADRP and also may be applicable to other inherited diseases.


Asunto(s)
Células Fotorreceptoras/patología , Mutación Puntual , ARN Catalítico/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Rodopsina/genética , Animales , Animales Modificados Genéticamente , Dependovirus , Modelos Animales de Enfermedad , Genes Dominantes , Terapia Genética , Histidina , Prolina , Regiones Promotoras Genéticas , ARN Catalítico/biosíntesis , ARN Catalítico/genética , Ratas , Ratas Sprague-Dawley , Células Fotorreceptoras Retinianas Bastones/metabolismo , Rodopsina/metabolismo , Opsinas de Bastones/genética
14.
Gene Ther ; 17(9): 1162-74, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20428215

RESUMEN

A prerequisite for using corrective gene therapy to treat humans with inherited retinal degenerative diseases that primarily affect rods is to develop viral vectors that target specifically this population of photoreceptors. The delivery of a viral vector with photoreceptor tropism coupled with a rod-specific promoter is likely to be the safest and most efficient approach to target expression of the therapeutic gene to rods. Three promoters that included a fragment of the proximal mouse opsin promoter (mOP), the human G-protein-coupled receptor protein kinase 1 promoter (hGRK1), or the cytomegalovirus immediate early enhancer combined with the chicken ß actin proximal promoter CBA were evaluated for their specificity and robustness in driving GFP reporter gene expression in rods, when packaged in a recombinant adeno-associated viral vector of serotype 2/5 (AAV2/5), and delivered via subretinal injection to the normal canine retina. Photoreceptor-specific promoters (mOP, hGRK1) targeted robust GFP expression to rods, whereas the ubiquitously expressed CBA promoter led to transgene expression in the retinal pigment epithelium, rods, cones and rare Müller, horizontal and ganglion cells. Late onset inflammation was frequently observed both clinically and histologically with all three constructs when the highest viral titers were injected. Cone loss in the injected regions of the retinas that received the highest titers occurred with both the hGRK1 and CBA promoters. Efficient and specific rod transduction, together with preservation of retinal structure was achieved with both mOP and hGRK1 promoters when viral titers in the order of 10(11)vg ml(-1) were used.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Regiones Promotoras Genéticas , Células Fotorreceptoras Retinianas Bastones/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Perros , Quinasas de Receptores Acoplados a Proteína-G/genética , Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Genes Reporteros/genética , Vectores Genéticos/genética , Humanos , Ratones , Transfección
15.
Gene Ther ; 17(7): 815-26, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20237510

RESUMEN

To test whether fast-acting, self-complimentary (sc), adeno-associated virus-mediated RPE65 expression prevents cone degeneration and/or restores cone function, we studied two mouse lines: the Rpe65-deficient rd12 mouse and the Rpe65-deficient, rhodopsin null ('that is, cone function-only') Rpe65(-/-)::Rho(-/-) mouse. scAAV5 expressing RPE65 was injected subretinally into one eye of rd12 and Rpe65(-/-)::Rho(-/-) mice at postnatal day 14 (P14). Contralateral rd12 eyes were injected later, at P35. Rd12 behavioral testing revealed that rod vision loss was prevented with either P14 or P35 treatment, whereas cone vision was only detected after P14 treatment. Consistent with this observation, P35 treatment only restored rod electroretinogram (ERG) signals, a result likely due to reduced cone densities at this time point. For Rpe65(-/-)::Rho(-/-) mice in which there is no confounding rod contribution to the ERG signal, cone cells and cone-mediated ERGs were also maintained with treatment at P14. This work establishes that a self-complimentary AAV5 vector can restore substantial visual function in two genetically distinct models of Rpe65 deficiency within 4 days of treatment. In addition, this therapy prevents cone degeneration but only if administered before extensive cone degeneration, thus supporting continuation of current Leber's congenital amaurosis-2 clinical trials with an added emphasis on cone subtype analysis and early intervention.


Asunto(s)
Proteínas Portadoras/fisiología , Dependovirus/genética , Proteínas del Ojo/fisiología , Terapia Genética , Atrofia Óptica Hereditaria de Leber/terapia , Células Fotorreceptoras Retinianas Conos/fisiología , Degeneración Retiniana/genética , Degeneración Retiniana/prevención & control , Animales , Modelos Animales de Enfermedad , Vectores Genéticos , Ratones , cis-trans-Isomerasas
16.
Gene Ther ; 16(1): 10-6, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18633446

RESUMEN

Vascular endothelial growth factor (VEGF) is important in pathological neovascularization, which is a key component of diseases such as the wet form of age-related macular degeneration, proliferative diabetic retinopathy and cancer. One of the most potent naturally occurring VEGF binders is VEGF receptor Flt-1. We have generated two novel chimeric VEGF-binding molecules, sFLT01 and sFLT02, which consist of the second immunoglobulin (IgG)-like domain of Flt-1 fused either to a human IgG1 Fc or solely to the CH3 domain of IgG1 Fc through a polyglycine linker 9Gly. In vitro analysis showed that these novel molecules are high-affinity VEGF binders. We have demonstrated that adeno-associated virus serotype 2 (AAV2)-mediated intravitreal gene delivery of sFLT01 efficiently inhibits angiogenesis in the mouse oxygen-induced retinopathy model. There were no histological observations of toxicity upon persistent ocular expression of sFLT01 for up to 12 months following intravitreal AAV2-based delivery in the rodent eye. Our data suggest that AAV2-mediated intravitreal gene delivery of our novel molecules may be a safe and effective treatment for retinal neovascularization.


Asunto(s)
Terapia Genética/métodos , Proteínas Recombinantes de Fusión/genética , Retina/metabolismo , Neovascularización Retiniana/terapia , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Dependovirus/genética , Técnica del Anticuerpo Fluorescente , Expresión Génica , Ingeniería Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/análisis , Proteínas Recombinantes de Fusión/metabolismo , Neovascularización Retiniana/metabolismo , Transducción Genética/métodos , Transgenes , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
17.
Mol Vis ; 15: 1835-42, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19756181

RESUMEN

PURPOSE: To compare self-complementary (sc) and single-stranded (ss) adeno-associated viral 2/5 (AAV2/5) vectors for retinal cell transduction in the dog when delivered by subretinal injection. METHODS: ScAAV2/5 and ssAAV2/5 vectors encoding enhanced green fluorescent protein (GFP) under control of the chicken beta actin promoter were prepared to the same titer. Equal amounts of viral particles were delivered into the subretinal spaces of both eyes of two dogs. In each dog, one eye received the scAAV2/5 and the other the ssAAV2/5. In vivo expression of GFP was monitored ophthalmoscopically. The dogs were sacrificed, and their retinas were examined by fluorescent microscopy and immunohistochemistry to determine GFP expression patterns and to assay for glial reactivity. RESULTS: GFP expression in the scAAV2/5 injected eyes was detectable at a much earlier time point than in the ssAAV2/5 injected eyes. Expression of GFP was also at higher levels in the scAAV2/5-injected eyes. Expression levels remained stable for the seven month duration of the study. The types of cells transduced by both vectors were similar; there was strong reporter gene expression in the RPE and photoreceptors, although not all cones in the transduced area expressed GFP. Some horizontal and Müller cells were also transduced. CONCLUSIONS: When delivered by subretinal injection in the dog, scAAV2/5 induces faster and stronger transgene expression than ssAAV2/5. The spectrum of retinal neurons transduced is similar between the two vectors. These results confirm in a large animal model those previously reported in the mouse. ScAAV2/5 shows promise for use in the treatment of conditions where a rapid transgene expression is desirable. Furthermore, it may be possible to use a lower number of viral particles to achieve the same effect compared with ssAAV2/5 vectors.


Asunto(s)
ADN Complementario/genética , Dependovirus/genética , Vectores Genéticos/genética , Transducción Genética , Animales , Perros , Femenino , Proteína Ácida Fibrilar de la Glía , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Masculino , Modelos Animales , Neuroglía/citología , Retina/citología
18.
Gene Ther ; 15(14): 1049-55, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18337838

RESUMEN

Specific cone-directed therapy is of high priority in the treatment of human hereditary retinal diseases. However, not much information exists about the specific targeting of photoreceptor subclasses. Three versions of the human red cone opsin promoter (PR0.5, 3LCR-PR0.5 and PR2.1), and the human blue cone opsin promoter HB569, were evaluated for their specificity and robustness in targeting green fluorescent protein (GFP) gene expression to subclasses of cones in the canine retina when used in recombinant adeno-associated viral vectors of serotype 5. The vectors were administered by subretinal injection. The promoter PR2.1 led to most effective and specific expression of GFP in the long- and medium-wavelength-absorbing cones (L/M cones) of normal and diseased retinas. The PR0.5 promoter was not effective. Adding three copies of the 35-bp LCR in front of PR0.5 lead to weak GFP expression in L/M cones. The HB569 promoter was not specific, and GFP was expressed in a few L/M cones, some rods and the retinal pigment epithelium. These results suggest that L/M cones, the predominant class of cone photoreceptors in the retinas of dogs and most mammalian species can be successfully targeted using the human red cone opsin promoter.


Asunto(s)
Terapia Genética/métodos , Regiones Promotoras Genéticas , Células Fotorreceptoras Retinianas Conos/metabolismo , Opsinas de Bastones/genética , Reparación del Gen Blanco , Animales , Defectos de la Visión Cromática/metabolismo , Defectos de la Visión Cromática/terapia , Dependovirus/genética , Perros , Expresión Génica , Vectores Genéticos/administración & dosificación , Proteínas Fluorescentes Verdes/genética , Humanos , Inyecciones , Modelos Animales , Transducción Genética/métodos , Transgenes
19.
Mol Vis ; 14: 2087-96, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19023450

RESUMEN

PURPOSE: Diseased corneas are potential targets for viral-based gene therapy to normalize (stimulate or inhibit) the expression of specific proteins. The choice of viral vectors is important to achieve optimal effect. The purpose of this study was to compare the tropism to different corneal cells of recombinant adenovirus (rAV) and recombinant adeno-associated virus (rAAV) constructs using live rabbit and organ-cultured human corneas. METHODS: rAV constructs harbored the green fluorescent protein (GFP) gene under the control of major immediate early cytomegalovirus (CMV) promoter. rAAV constructs from virus serotypes 1, 2 5, 7, and 8 had GFP under the chicken beta-actin promoter and CMV enhancer. For organ culture, 16 healthy and diabetic postmortem human corneas were used. Five or fifteen microl rAV at 10(7) plaque forming units per 1 microl were added for 2 days to culture medium of uninjured corneas that were further cultured for 5-32 days. rAAV were added at 1.2-7.8x10(10) vector genomes per cornea for 3 days to each cornea; the culture then continued for another 14-23 days. Corneal cryostat sections were examined by immunohistochemistry. Live rabbit corneas were used following excimer laser ablation of the corneal epithelium with preservation of the basal cell layer. Equal numbers of rAAV particles (2x10(11) vector genomes) were applied to the cornea for 10 min. After seven days to allow for corneal healing and gene expression the animals were euthanized, the corneas were excised, and sections analyzed by immunohistochemistry. RESULTS: By direct fluorescence microscopy of live organ-cultured human corneas GFP signal after rAV transduction was strong in the epithelium with dose-dependent intensity. On corneal sections, GFP was seen in all epithelial layers and some endothelial cells but most keratocytes were negative. In rAAV-transduced organ-cultured human corneas GFP signal could only be detected with anti-GFP antibody immunohistochemistry. GFP was observed in the epithelium, keratocytes, and endothelium, with more pronounced basal epithelial cell staining with rAAV1 than with other serotypes. No difference in the GFP expression patterns or levels between normal and diabetic corneas was noted. The rabbit corneas showed very similar patterns of GFP distribution to human corneas. With all rAAV serotype vectors, GFP staining in the epithelium was significantly (p=0.007) higher than the background staining in non-transduced corneas, with a trend for rAAV1 and rAAV8 to produce higher staining intensities than for rAAV2, rAAV5 (p=0.03; rAAV5 versus rAAV1), and rAAV7. rAAV serotype vectors also transduced stromal and endothelial cells in rabbit corneas to a different extent. CONCLUSIONS: rAAV appears to reach many more corneal cells than rAV, especially keratocytes, although GFP expression levels were lower compared to rAV. rAV may be more useful than rAAV for gene therapy applications requiring high protein expression levels, but rAAV may be superior for keratocyte targeting.


Asunto(s)
Adenoviridae/metabolismo , Córnea/citología , Córnea/metabolismo , Dependovirus/metabolismo , Terapia Genética , Animales , Pollos , Córnea/patología , Diabetes Mellitus/patología , Epitelio Corneal/citología , Vectores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Técnicas de Cultivo de Órganos , Especificidad de Órganos , Conejos , Transducción Genética
20.
Mol Cell Biol ; 13(4): 2162-71, 1993 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8455604

RESUMEN

A methylation protection assay was used in a novel manner to demonstrate a specific bovine protein-mitochondrial DNA (mtDNA) interaction within the organelle (in organello). The protected domain, located near the D-loop 3' end, encompasses a conserved termination-associated sequence (TAS) element which is thought to be involved in the regulation of mtDNA synthesis. In vitro footprinting studies using a bovine mitochondrial extract and a series of deleted mtDNA templates identified a approximately 48-kDa protein which binds specifically to a single TAS element also protected within the mitochondrion. Because other TAS-like elements located in close proximity to the protected region did not footprint, protein binding appears to be highly sequence specific. The in organello and in vitro data, together, provide evidence that D-loop formation is likely to be mediated, at least in part, through a trans-acting factor binding to a conserved sequence element located 58 bp upstream of the D-loop 3' end.


Asunto(s)
ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/metabolismo , Mitocondrias/química , Animales , Secuencia de Bases , Sitios de Unión , Química Encefálica , Bovinos , Clonación Molecular , Reactivos de Enlaces Cruzados , ADN Mitocondrial/química , Desoxirribonucleasa I/farmacología , Metilación , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA